Viral activation of MK2-hsp27-p115RhoGEF-RhoA signaling axis causes cytoskeletal rearrangements, p-body disruption and ARE-mRNA stabilization

PLoS Pathog. 2015 Jan 8;11(1):e1004597. doi: 10.1371/journal.ppat.1004597. eCollection 2015 Jan.

Abstract

Kaposi's sarcoma-associated herpesvirus (KSHV) is the infectious cause of several AIDS-related cancers, including the endothelial cell (EC) neoplasm Kaposi's sarcoma (KS). KSHV-infected ECs secrete abundant host-derived pro-inflammatory molecules and angiogenic factors that contribute to tumorigenesis. The precise contributions of viral gene products to this secretory phenotype remain to be elucidated, but there is emerging evidence for post-transcriptional regulation. The Kaposin B (KapB) protein is thought to contribute to the secretory phenotype in infected cells by binding and activating the stress-responsive kinase MK2, thereby selectively blocking decay of AU-rich mRNAs (ARE-mRNAs) encoding pro-inflammatory cytokines and angiogenic factors. Processing bodies (PBs) are cytoplasmic ribonucleoprotein foci in which ARE-mRNAs normally undergo rapid 5' to 3' decay. Here, we demonstrate that PB dispersion is a feature of latent KSHV infection, which is dependent on kaposin protein expression. KapB is sufficient to disperse PBs, and KapB-mediated ARE-mRNA stabilization could be partially reversed by treatments that restore PBs. Using a combination of genetic and chemical approaches we provide evidence that KapB-mediated PB dispersion is dependent on activation of a non-canonical Rho-GTPase signaling axis involving MK2, hsp27, p115RhoGEF and RhoA. PB dispersion in latently infected cells is likewise dependent on p115RhoGEF. In addition to PB dispersion, KapB-mediated RhoA activation in primary ECs caused actin stress fiber formation, increased cell motility and angiogenesis; these effects were dependent on the activity of the RhoA substrate kinases ROCK1/2. By contrast, KapB-mediated PB dispersion occurred in a ROCK1/2-independent manner. Taken together, these observations position KapB as a key contributor to viral reprogramming of ECs, capable of eliciting many of the phenotypes characteristic of KS tumor cells, and strongly contributing to the post-transcriptional control of EC gene expression and secretion.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • AU Rich Elements / genetics
  • Cells, Cultured
  • Cytoplasmic Vesicles / metabolism*
  • Cytoskeleton / metabolism*
  • HEK293 Cells
  • HSP27 Heat-Shock Proteins / metabolism
  • HeLa Cells
  • Heat-Shock Proteins
  • Herpesvirus 8, Human / physiology*
  • Human Umbilical Vein Endothelial Cells
  • Humans
  • Intracellular Signaling Peptides and Proteins / metabolism
  • Molecular Chaperones
  • Protein Serine-Threonine Kinases / metabolism
  • RNA Processing, Post-Transcriptional
  • RNA Stability / physiology*
  • Rho Guanine Nucleotide Exchange Factors / metabolism
  • Sarcoma, Kaposi / virology
  • Signal Transduction / physiology
  • rhoA GTP-Binding Protein / metabolism

Substances

  • ARHGEF1 protein, human
  • HSP27 Heat-Shock Proteins
  • HSPB1 protein, human
  • Heat-Shock Proteins
  • Intracellular Signaling Peptides and Proteins
  • Molecular Chaperones
  • Rho Guanine Nucleotide Exchange Factors
  • MAP-kinase-activated kinase 2
  • Protein Serine-Threonine Kinases
  • rhoA GTP-Binding Protein